WO2020030927A1 - Peptides de pénétration cellulaire - Google Patents

Peptides de pénétration cellulaire Download PDF

Info

Publication number
WO2020030927A1
WO2020030927A1 PCT/GB2019/052247 GB2019052247W WO2020030927A1 WO 2020030927 A1 WO2020030927 A1 WO 2020030927A1 GB 2019052247 W GB2019052247 W GB 2019052247W WO 2020030927 A1 WO2020030927 A1 WO 2020030927A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
peptide
suitably
cationic
hyp
Prior art date
Application number
PCT/GB2019/052247
Other languages
English (en)
Inventor
Matthew Wood
Raquel MANZANO
Caroline GODFREY
Graham Mcclorey
Richard Raz
Michael Gait
Andrey ARZUMANOV
Liz O'DONOVAN
Gareth HAZELL
Ashling HOLLAND
Miguel VARELA
Original Assignee
Oxford University Innovation Limited
United Kingdom Research And Innovation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Oxford University Innovation Limited, United Kingdom Research And Innovation filed Critical Oxford University Innovation Limited
Priority to AU2019317789A priority Critical patent/AU2019317789A1/en
Priority to KR1020217007003A priority patent/KR20210090157A/ko
Priority to CA3108876A priority patent/CA3108876A1/fr
Priority to JP2021506986A priority patent/JP2021534131A/ja
Priority to EP19755420.7A priority patent/EP3833376A1/fr
Priority to MX2021001545A priority patent/MX2021001545A/es
Priority to BR112021002447-6A priority patent/BR112021002447A2/pt
Priority to CN201980066823.5A priority patent/CN113347989A/zh
Priority to US17/266,939 priority patent/US20210299263A1/en
Publication of WO2020030927A1 publication Critical patent/WO2020030927A1/fr
Priority to IL280678A priority patent/IL280678A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • A61K47/6455Polycationic oligopeptides, polypeptides or polyamino acids, e.g. for complexing nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/10Fusion polypeptide containing a localisation/targetting motif containing a tag for extracellular membrane crossing, e.g. TAT or VP22
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Definitions

  • the present invention relates to peptides, in particular cell-penetrating peptides, and to conjugates of such cell-penetrating peptides with a therapeutic molecule.
  • the present invention further relates to use of such peptides or conjugates in methods of treatment or as a medicament, especially in the treatment of genetic disorders and in particular muscular dystrophies such as Duchenne muscular dystrophy.
  • Nucleic acid drugs are genomic medicines with the potential to transform human healthcare. Research has indicated that such therapeutics could have applications across a broad range of disease areas including neuromuscular disease.
  • Duchenne muscular dystrophy (DMD) has placed this monogenic disorder at the forefront of advances in precision medicine.
  • DMD affects one in 3500 new born boys. This severe, X-linked recessive disease results from mutations in the DMD gene that encodes dystrophin protein.
  • the disorder is characterised by progressive muscle degeneration and wasting, along with the emergence of respiratory failure and cardiac complications, ultimately leading to premature death.
  • the majority of mutations underlying DMD are genomic out-of-frame deletions that induce a premature truncation in the open reading frame that results in the absence of the dystrophin protein.
  • Exon skipping therapy utilises splice switching antisense oligonucleotides (SSOs) to target specific regions of the DMD transcript, inducing the exclusion of individual exons, leading to the restoration of aberrant reading frames and resulting in the production of an internally deleted, yet partially functional, dystrophin protein.
  • SSOs splice switching antisense oligonucleotides
  • viruses as delivery vehicles has been suggested, however their use is limited due to the immunotoxicity of the viral coat protein and potential oncogenic effects.
  • a range of non-viral delivery vectors have been developed, amongst which peptides have shown the most promise due to their small size, targeting specificity and ability of trans-capillary delivery of large bio-cargoes.
  • Several peptides have been reported for their ability to permeate cells either alone or carrying a bio-cargo.
  • CPPs cell-penetrating peptides
  • SSOs charge neutral phosphorodiamidate morpholino oligomers (PMO) and peptide nucleic acids (PNA)
  • PMO charge neutral phosphorodiamidate morpholino oligomers
  • PNA peptide nucleic acids
  • PNA/PMO internalization peptides have been developed which are arginine-rich CPPs that are comprised of two arginine-rich sequences separated by a central short hydrophobic sequence. These‘Pip’ peptides were designed to improve serum stability whilst maintaining a high level of exon skipping, initially by attachment to a PNA cargo. Further derivatives of these peptides were designed as conjugates of PMOs, which were shown to lead to body-wide skeletal muscle dystrophin production, and importantly also including the heart, following systemic administration in mice. Despite these peptides being efficacious, their therapeutic application has been hampered by their associated toxicity.
  • the challenge in the field of cell-penetrating peptide technology has been to de-couple efficacy and toxicity.
  • the present inventors have now identified, synthesized and tested a number of improved CPPs having a particular structure according to the present invention which address at least this problem.
  • peptides maintain good levels of efficacy in skeletal muscles when tested in vitro and in vivo with a cargo therapeutic molecule. Furthermore, these peptides demonstrate an improvement in efficacy compared with previously available CPPs when used in the same conjugate. At the same time, these peptides act effectively in vivo with reduced clinical signs following systemic injection and lower toxicity as observed through measurement of biochemical markers. Crucially, the present peptides are demonstrated to show a surprisingly reduced toxicity following similar systemic injection into mice when compared with previous CPPs. Accordingly, the peptides of the invention offer improved suitability for use as a therapy for humans than previously available peptides and can be used in therapeutic conjugates for safe and effective treatment of human subjects.
  • a peptide having a total length of 40 amino acid residues or less comprising: two or more cationic domains each comprising at least 4 amino acid residues; and one or more hydrophobic domains each comprising at least 3 amino acid residues; wherein the peptide does not contain artificial amino acid residues.
  • a conjugate comprising the peptide of the first aspect covalently linked to a therapeutic molecule.
  • a conjugate comprising the peptide of the first aspect covalently linked to an imaging molecule.
  • a pharmaceutical composition comprising the conjugate of the second aspect.
  • a conjugate according to the second aspect for use as a medicament.
  • a pharmaceutical composition according to the fourth aspect for use as a medicament.
  • a method of treating a disease in a subject comprising administering the conjugate of the second aspect to the subject in a therapeutically effective amount.
  • a method of treating a disease in a subject comprising administering the pharmaceutical composition according to the fourth aspect to the subject in a therapeutically effective amount.
  • an isolated nucleic acid encoding the peptide of the first aspect or the conjugate of the second aspect or the conjugate of the third aspect.
  • an expression vector comprising the nucleic acid sequence of the seventh aspect.
  • a host cell comprising the expression vector of the eighth aspect.
  • the inventors have produced a series of peptides that are suitable for use as cell-penetrating peptides to deliver therapeutic molecules into cells.
  • the inventors have discovered a group of peptides having at least two cationic domains and at least one hydrophobic domain of defined lengths without any artificial amino acids which, provide enhanced cell penetration into muscles, compared with currently available cell-penetrating peptides. This effect is observed when delivered as a conjugate with an antisense oligonucleotide therapeutic into cells, or when administered in vivo.
  • enhanced cell penetration by peptides of the invention linked to a suitable therapeutic molecule can be shown by specific exon exclusion within the transcript.
  • the directing of an antisense oligonucleotide to an appropriate sequence results in the forced skipping of an exon, the correction of the open reading frame and the restoration of an internally deleted, yet partially functional isoform of dystrophin.
  • the peptides of the present invention when used as a conjugate with an antisense oligonucleotide therapeutic designed to target the dystrophin gene are shown herein to have high levels of exon exclusion and dystrophin protein restoration.
  • the conjugates comprising peptides of the invention show significantly increased cell penetration when compared with currently available peptides conjugated to the same antisense oligonucleotide therapeutic. This is demonstrated in the present invention by increased exon skipping in the dystrophin gene in various different muscle groups.
  • results described herein show levels exon skipping and functional dystrophin expression when using peptide conjugates of the invention approaching double the levels resulting from the use of the same antisense oligonucleotide therapeutic conjugated to a previously available cell-penetrating peptides.
  • peptide structure that does not contain any artificial amino acid residues would increase the delivery properties of previously reported cell-penetrating peptides to transport a therapeutic molecule cargo, such as an oligonucleotide, into muscle.
  • a peptide’s effectiveness largely depends on its ability to be serum-stable for the required length of time for it to get into cells. It was expected that peptides formed without artificial amino acids would be too unstable and vulnerable to protease degradation in vivo to allow a sufficient amount to penetrate muscle cells and tissue and result in enhanced therapeutic effect. Contrary to this expectation, the inventors found that peptides having a particular structure as claimed are stable enough to enter cells and maintain good, even improved efficacy, yet have the advantage of a reduced toxicity profile due to lacking artificial amino acids.
  • the invention includes any combination of the aspects and features described except where such a combination is clearly impermissible or expressly avoided.
  • the present invention relates to short cell-penetrating peptides that have a particular structure in which there are no artificial amino acid residues.
  • references to an‘artificial’ amino acid or residue herein denotes any amino acid that does not occur in nature and includes synthetic amino acids, modified amino acids (such as those modified with sugars), non-natural amino acids, man-made amino acids, spacers, and non peptide bonded spacers.
  • Synthetic amino acids may be those that are chemically synthesised by man.
  • aminohexanoic acid is an artificial amino acid in the context of the present invention.
  • beta-alanine B
  • hydroxyproline Hyp
  • Artificial amino acids may include, for example, 6-aminohexanoic acid (X), tetrahydroisoquinoline-3-carboxylic acid (TIC), 1-(amino)cyclohexanecarboxylic acid (Cy), and 3-azetidine-carboxylic acid (Az), 11-aminoundecanoic acid.
  • X 6-aminohexanoic acid
  • TIC tetrahydroisoquinoline-3-carboxylic acid
  • Cy 1-(amino)cyclohexanecarboxylic acid
  • Az 3-azetidine-carboxylic acid
  • the peptide does not contain aminohexanoic acid residues.
  • the peptide does not contain any form of aminohexanoic acid residues.
  • the peptide does not contain 6-aminohexanoic acid residues.
  • the peptide contains only natural amino acid residues, and therefore consists of natural amino acid residues.
  • artificial amino acids such as 6-aminohexanoic acid that are typically used in cell- penetrating peptides are replaced by natural amino acids.
  • artificial amino acids such as 6-aminohexanoic acid that are typically used in cell-penetrating peptides are replaced by amino acids selected from beta-alanine, serine, proline, arginine and histidine or hydroxyproline.
  • aminohexanoic acid is replaced by beta-alanine.
  • 6- aminohexanoic acid is replaced by beta-alanine
  • aminohexanoic acid is replaced by histidine.
  • 6-aminohexanoic acid is replaced by histidine.
  • aminohexanoic acid is replaced by hydroxyproline.
  • 6- aminohexanoic acid is replaced by hydroxyproline.
  • the artificial amino acids such as 6-aminohexanoic acid that are typically used in cell- penetrating peptides may be replaced by a combination of any of beta-alanine, serine, proline, arginine and histidine or hydroxyproline, suitably a combination of any of beta-alanine, histidine, and hydroxyproline.
  • a peptide having a total length of 40 amino acid residues or less comprising: two or more cationic domains each comprising at least 4 amino acid residues; and one or more hydrophobic domains each comprising at least 3 amino acid residues; wherein at least one cationic domain comprises histidine residues.
  • at least one cationic domain is histidine rich.
  • histidine rich is defined herein in relation to the cationic domains.
  • the present invention relates to short cell-penetrating peptides having a particular structure in which there are at least two cationic domains having a certain length.
  • references to‘cationic’ herein denote an amino acid or domain of amino acids having an overall positive charge at physiological pH.
  • the peptide comprises up to 4 cationic domains, up to 3 cationic domains.
  • the peptide comprises 2 cationic domains.
  • the peptide comprises two or more cationic domains each having a length of at least 4 amino acid residues.
  • each cationic domain has a length of between 4 to 12 amino acid residues, suitably a length of between 4 to 7 amino acid residues.
  • each cationic domain has a length of 4, 5, 6, or 7 amino acid residues.
  • each cationic domain is of similar length, suitably each cationic domain is the same length.
  • each cationic domain comprises cationic amino acids and may also contain polar and or nonpolar amino acids.
  • Non-polar amino acids may be selected from: alanine, beta-alanine, proline, glycine, cysteine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine.
  • Suitably non-polar amino acids do not have a charge.
  • Polar amino acids may be selected from: serine, asparagine, hydroxyproline, histidine, arginine, threonine, tyrosine, glutamine.
  • the selected polar amino acids do not have a negative charge.
  • Cationic amino acids may be selected from: arginine, histidine, lysine.
  • cationic amino acids have a positive charge at physiological pH.
  • each cationic domain does not comprise anionic or negatively charged amino acid residues.
  • each cationic domain comprises arginine, histidine, beta-alanine, hydroxyproline and/or serine residues.
  • each cationic domain consists of arginine, histidine, beta-alanine, hydroxyproline and/or serine residues.
  • each cationic domain comprises at least 40%, at least 45%, at least 50% cationic amino acids.
  • each cationic domain comprises a majority of cationic amino acids.
  • each cationic domain comprises at least 55%, at least 60%, at least 65% at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% cationic amino acids.
  • each cationic domain comprises an isoelectric point (pi) of at least 7.5, at least 8.0, at least 8.5, at least 9.0, at least 9.5, at least 10.0, at least 10.5, at least 11.0, at least 11.5, at least 12.0.
  • each cationic domain comprises an isoelectric point (pi) of at least 10.0.
  • each cationic domain comprises an isoelectric point (pi) of between 10.0 and 13.0
  • each cationic domain comprises an isoelectric point (pi) of between 10.4 and 12.5.
  • the isoelectric point of a cationic domain is calculated at physiological pH by any suitable means available in the art.
  • I PC www.isoelectric.org
  • each cationic domain comprises at least 1 cationic amino acid, suitably between 1-5 cationic amino acids.
  • each cationic domain comprises at least 2 cationic amino acids, suitably between 2-5 cationic amino acids.
  • each cationic domain is arginine rich and/or histidine rich.
  • a cationic domain may contain both histidine and arginine.
  • each cationic domain comprises a majority of arginine and/or histidine residues.
  • each cationic domain comprises at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 60%, at least 65%, least 70% arginine and/or histidine residues.
  • a cationic domain may comprise at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 60%, at least 65%, least 70% arginine residues.
  • a cationic domain may comprise at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 60%, at least 65%, least 70% histidine residues.
  • a cationic domain may comprise a total of between 1-5 histidine and 1-5 arginine residues.
  • a cationic domain may comprise between 1-5 arginine residues.
  • a cationic domain may comprise between 1-5 histidine residues.
  • a cationic domain may comprise a total of between 2-5 histidine and 3-5 arginine residues.
  • a cationic domain may comprise between 3-5 arginine residues.
  • a cationic domain may comprise between 2-5 histidine residues.
  • each cationic domain comprises one or more beta-alanine residues.
  • each cationic domain may comprise a total of between 2-5 beta-alanine residues, suitably a total of 2 or 3 beta-alanine residues.
  • a cationic domain may comprise one or more hydroxyproline residues or serine residues.
  • a cationic domain may comprise between 1-2 hydroxyproline residues.
  • a cationic domain may comprise between 1-2 serine residues.
  • cationic amino acids in a given cationic domain may be histidine, alternatively, suitably all of the cationic amino acids in a given cationic domain may be arginine.
  • the peptide may comprise at least one histidine rich cationic domain.
  • the peptide may comprise at least one arginine rich cationic domain.
  • the peptide may comprise at least one arginine rich cationic domain and at least one histidine rich cationic domain.
  • the peptide comprises two arginine rich cationic domains.
  • the peptide comprises two histidine rich cationic domains.
  • the peptide comprises two arginine and histidine rich cationic domains.
  • the peptide comprises one arginine rich cationic domain and one histidine rich cationic domain.
  • each cationic domain comprises no more than 3 contiguous arginine residues, suitably no more than 2 contiguous arginine residues.
  • each cationic domain comprises no contiguous histidine residues.
  • each cationic domain comprises arginine, histidine and/or beta-alanine residues.
  • each cationic domain comprises a majority of arginine, histidine and/or beta-alanine residues.
  • at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, 100% of the amino acid residues in each cationic domain are arginine, histidine and/or beta-alanine residues.
  • each cationic domain consists of arginine, histidine and/or beta-alanine residues.
  • the peptide comprises a first cationic domain comprising arginine and beta-alanine residues and a second cationic domain comprising arginine and beta-alanine residues.
  • the peptide comprises a first cationic domain comprising arginine and beta-alanine resides, and a second cationic domain comprising histidine, beta-alanine, and optionally arginine residues.
  • the peptide comprises a first cationic domain comprising arginine and beta-alanine resides, and a second cationic domain comprising histidine and beta-alanine residues.
  • the peptide comprises a first cationic domain consisting of arginine and beta-alanine residues and a second cationic domain consisting of arginine and beta-alanine residues.
  • the peptide comprises a first cationic domain consisting of arginine and beta-alanine residues and a second cationic domain consisting of arginine, histidine and beta- alanine residues.
  • the peptide comprises at least two cationic domains, suitably these cationic domains form the arms of the peptide.
  • the cationic domains are located at the N and C terminus of the peptide.
  • the cationic domains may be known as the cationic arm domains.
  • the peptide comprises two cationic domains, wherein one is located at the N-terminus of the peptide and one is located at the C-terminus of the peptide. Suitably at either end of the peptide. Suitably no further amino acids or domains are present at the N- terminus and C-terminus of the peptide, with the exception of other groups such as a terminal modification, linker and/or therapeutic molecule. For the avoidance of doubt, such other groups may be present in addition to‘the peptide’ described and claimed herein. Suitably therefore each cationic domain forms the terminus of the peptide. Suitably, this does not preclude the presence of a further linker group as described herein.
  • the peptide may comprise up to 4 cationic domains.
  • the peptide comprises two cationic domains.
  • the peptide comprises two cationic domains that are both arginine rich.
  • the peptide comprises one cationic domain that is arginine rich.
  • the peptide comprises two cationic domains that are both arginine and histidine rich.
  • the peptide comprises one cationic domain that is arginine rich and one cationic domain that is histidine rich.
  • the cationic domains comprise amino acid units selected from the following: R, H, B, RR, HH, BB, RH, HR, RB, BR, HB, BH, RBR, RBB, BRR, BBR, BRB, RBH, RHB, HRB, BRH, HRR, RRH, HRH, HBB, BBH, RHR, BHB, HBH, or any combination thereof.
  • a cationic domain may also include serine, proline and/or hydroxyproline residues.
  • the cationic domains may further comprise amino acid units selected from the following: RP, PR, RPR, RRP, PRR, PRP, Hyp; R[Hyp]R, RR[Hyp], [Hyp]RR, [Hyp]R[Hyp], [Hyp][Hyp]R, R[Hyp][Hyp], SB, BS, or any combination thereof, or any combination with the above listed amino acid units.
  • each cationic domain comprises any of the following sequences: RBRRBRR (SEQ ID NO: 1), RBRBR (SEQ ID NO:2), RBRR (SEQ ID NO:3), RBRRBR (SEQ ID NO:4), RRBRBR (SEQ ID NO:5), RBRRB (SEQ ID NO:6), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8), HBHBR (SEQ ID NO:9), RBRHBHR (SEQ ID NO:10), RBRBBHR (SEQ ID NO:1 1), RBRRBH (SEQ ID NO: 12), HBRRBR (SEQ ID NO: 13), HBHBH (SEQ ID NO: 14), BHBH (SEQ ID NO: 15), BRBSB (SEQ ID NO: 16), BRB[Hyp]B (SEQ ID NO: 17), R[Hyp]H[Hyp]HB (SEQ ID NO: 18), R[Hyp]RR[Hyp]R (SEQ ID NO:
  • each cationic domain consists any of the following sequences: RBRRBRR (SEQ ID NO: 1), RBRBR (SEQ ID NO:2), RBRR (SEQ ID NO:3), RBRRBR (SEQ ID NO:4), RRBRBR (SEQ ID NO:5), RBRRB (SEQ ID NO:6), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8), HBHBR (SEQ ID NO:9), RBRHBHR (SEQ ID NO:10), RBRBBHR (SEQ ID NO:1 1), RBRRBH (SEQ ID NO: 12), HBRRBR (SEQ ID NO:13) , HBHBH (SEQ ID NO: 14), BHBH (SEQ ID NO:15), BRBSB (SEQ ID NO:16), BRB[Hyp]B, R[Hyp]H[Hyp]HB, R[Hyp]RR[Hyp]R (SEQ ID NO: 19) or any combination thereof.
  • each cationic domain consists of one of the following sequences: RBRRBRR (SEQ ID NO:1), RBRBR (SEQ ID NO:2), RBRRBR (SEQ ID NO:4), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8), HBHBR (SEQ ID NO:9).
  • each cationic domain in the peptide may be identical or different.
  • each cationic domain in the peptide is different.
  • the present invention relates to short cell-penetrating peptides having a particular structure in which there is at least one hydrophobic domain having a certain length.
  • references to‘hydrophobic’ herein denote an amino acid or domain of amino acids having the ability to repel water or which do not mix with water.
  • the peptide comprises up to 3 hydrophobic domains, up to 2 hydrophobic domains.
  • the peptide comprises 1 hydrophobic domain.
  • the peptide comprises one or more hydrophobic domains each having a length of at least 3 amino acid residues.
  • each hydrophobic domain has a length of between 3-6 amino acids.
  • each hydrophobic domain has a length of 5 amino acids.
  • each hydrophobic domain may comprise nonpolar, polar, and hydrophobic amino acid residues.
  • Hydrophobic amino acid residues may be selected from: alanine, valine, leucine, isoleucine, phenylalanine, tyrosine, methionine, and tryptophan.
  • Non-polar amino acid residues may be selected from: proline, glycine, cysteine, alanine, valine, leucine, isoleucine, tryptophan, phenylalanine, methionine.
  • Polar amino acid residues may be selected from: Serine, Asparagine, hydroxyproline, histidine, arginine, threonine, tyrosine, glutamine.
  • hydrophobic domains do not comprise hydrophilic amino acid residues.
  • each hydrophobic domain comprises a majority of hydrophobic amino acid residues.
  • each hydrophobic domain comprises at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, 100% hydrophobic amino acids.
  • each hydrophobic domain consists of hydrophobic amino acid residues.
  • each hydrophobic domain comprises a hydrophobicity of at least 0.3, at least 0.4, at least 0.5, at least 0.6, at least 0.7, at least 0.8, at least 0.8, at least 1.0, at least 1.1 , at least 1.2, at least 1.3.
  • each hydrophobic domain comprises a hydrophobicity of at least 0.3, at least 0.35, at least 0.4, at least 0.45.
  • each hydrophobic domain comprises a hydrophobicity of at least 1.2, at least 1.25, at least 1.3, at least 1.35.
  • each hydrophobic domain comprises a hydrophobicity of between 0.4 and 1.4
  • each hydrophobic domain comprises of a hydrophobicity of between 0.45 and 0.48.
  • each hydrophobic domain comprises a hydrophobicity of between 1.27 and 1.39
  • hydrophobicity is as measured by White and Wimley: W.C. Wimley and S.H. White, "Experimentally determined hydrophobicity scale for proteins at membrane interfaces” Nature Struct Biol 3:842 (1996).
  • each hydrophobic domain comprises at least 3, at least 4 hydrophobic amino acid residues.
  • each hydrophobic domain comprises phenylalanine, leucine, Isoleucine, tyrosine, tryptophan, proline, and glutamine residues.
  • each hydrophobic domain consists of phenylalanine, leucine, isoleucine, tyrosine, tryptophan, proline, and/or glutamine residues.
  • each hydrophobic domain consists of phenylalanine, leucine, isoleucine, tyrosine and/or glutamine residues.
  • each hydrophobic domain consists of tryptophan and/or proline residues.
  • the peptide comprises one hydrophobic domain.
  • the or each hydrophobic domain is located in the centre of the peptide.
  • the hydrophobic domain may be known as a core hydrophobic domain.
  • the or each hydrophobic core domain is flanked on either side by an arm domain.
  • the arm domains may comprise one or more cationic domains and one or more further hydrophobic domains.
  • each arm domain comprises a cationic domain.
  • the peptide comprises two arm domains flanking a hydrophobic core domain, wherein each arm domain comprises a cationic domain.
  • the peptide consists of two cationic arm domains flanking a hydrophobic core domain.
  • the or each hydrophobic domain comprises one of the following sequences: YQFLI (SEQ ID NO:20), FQILY (SEQ ID NO:21), ILFQY (SEQ ID NO:22), FQIY (SEQ ID NO:23), WWW, WWPWW (SEQ ID NO:24), WPWW (SEQ ID NO:25), WWPW (SEQ ID NO:26) or any combination thereof.
  • the or each hydrophobic domain consists of one of the following sequences: YQFLI (SEQ ID NO:20), FQILY (SEQ ID NO:21), ILFQY (SEQ ID NO:22), FQIY (SEQ ID NO:23), WWW, WWPWW (SEQ ID NO:24), WPWW (SEQ ID NO:25), WWPW (SEQ ID NO:26) or any combination thereof.
  • the or each hydrophobic domain consists of one of the following sequences FQILY (SEQ ID NO:21), YQFLI (SEQ ID NO:20), ILFQY (SEQ ID NO:22).
  • the or each hydrophobic domain consists of FQILY (SEQ ID NO:21).
  • each hydrophobic domain in the peptide may have the same sequence or a different sequence.
  • the present invention relates to short cell-penetrating peptides for use in transporting therapeutic cargo molecules in the treatment of medical conditions.
  • the peptide has a sequence that is a contiguous single molecule, therefore the domains of the peptide are contiguous.
  • the peptide comprises several domains in a linear arrangement between the N-terminus and the C-terminus.
  • the domains are selected from cationic domains and hydrophobic domains described above.
  • the peptide consists of cationic domains and hydrophobic domains wherein the domains are as defined above.
  • Each domain has common sequence characteristics as described in the relevant sections above, but the exact sequence of each domain is capable of variation and modification. Thus a range of sequences is possible for each domain.
  • the combination of each possible domain sequence yields a range of peptide structures, each of which form part of the present invention. Features of the peptide structures are described below.
  • a hydrophobic domain separates any two cationic domains.
  • each hydrophobic domain is flanked by cationic domains on either side thereof.
  • no cationic domain is contiguous with another cationic domain.
  • the peptide comprises one hydrophobic domain flanked by two cationic domains in the following arrangement:
  • the hydrophobic domain may be known as the core domain and each of the cationic domains may be known as an arm domain.
  • the hydrophobic arm domains flank the cationic core domain on either side thereof.
  • the peptide consists of two cationic domains and one hydrophobic domain.
  • the peptide consists of one hydrophobic core domain flanked by two cationic arm domains.
  • the peptide consists of one hydrophobic core domain comprising a sequence selected from: YQFLI (SEQ ID NO:20), FQILY (SEQ ID NO:21), ILFQY (SEQ ID NO:22), FQIY (SEQ ID NO:23), WWW, WWPWW (SEQ ID NO:24), WPWW (SEQ ID NO:25), and WWPW (SEQ ID NO:26), flanked by two cationic arm domains each comprising a sequence selected from: RBRRBRR (SEQ ID NO: 1), RBRBR (SEQ ID NO:2), RBRR (SEQ ID NO:3), RBRRBR (SEQ ID NO:4), RRBRBR (SEQ ID NO:5), RBRRB (SEQ ID NO:6), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8), HBHBR (SEQ ID NO:9), RBRHBHR (SEQ ID NO: 10), RBRBBHR (SEQ ID NO:20
  • the peptide consists of one hydrophobic core domain comprising a sequence selected from: FQILY (SEQ ID NO:21), YQFLI (SEQ ID NO:20), and ILFQY (SEQ ID NO:22), flanked by two cationic arm domains comprising a sequence selected from: RBRRBRR (SEQ ID NO: 1), RBRBR (SEQ ID NO:2), RBRRBR (SEQ ID NO:4), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8), HBHBR (SEQ ID NO:9).
  • FQILY SEQ ID NO:21
  • YQFLI SEQ ID NO:20
  • ILFQY SEQ ID NO:22
  • flanked by two cationic arm domains comprising a sequence selected from: RBRRBRR (SEQ ID NO: 1), RBRBR (SEQ ID NO:2), RBRRBR (SEQ ID NO:4), BRBR (SEQ ID NO:7), RBHBH (
  • the peptide consists of one hydrophobic core domain comprising the sequence: FQILY (SEQ ID NO:21), flanked by two cationic arm domains comprising a sequence selected from: RBRRBRR (SEQ ID NO:1), RBRBR (SEQ ID NO:2), RBRRBR (SEQ ID NO:4), BRBR (SEQ ID NO:7), RBHBH (SEQ ID NO:8).
  • further groups may be present such as a linker, terminal modification and/or therapeutic molecule.
  • the peptide is N-terminally modified.
  • the peptide is N-acetylated, N-methylated, N-trifluoroacetylated, N- trifluoromethylsulfonylated, or N-methylsulfonylated.
  • the peptide is N-acetylated.
  • the N-terminus of the peptide may be unmodified.
  • the peptide is N-acetylated.
  • the peptide is C-terminal modified.
  • the peptide comprises a C-terminal modification selected from: Carboxy-, Thioacid-, Aminooxy-, Hydrazino-, thioester-, azide, strained alkyne, strained alkene, aldehyde-, thiol or haloacetyl-group.
  • the C-terminal modification provides a means for linkage of the peptide to the therapeutic molecule.
  • the C-terminal modification may comprise the linker and vice versa.
  • the C-terminal modification may consist of the linker or vice versa. Suitable linkers are described herein elsewhere.
  • the peptide comprises a C-terminal carboxyl group.
  • the C-terminal carboxyl group is provided by a glycine or beta-alanine residue.
  • the C terminal carboxyl group is provided by a beta-alanine residue.
  • the C terminal beta-alanine residue is a linker.
  • each cationic domain may further comprise an N or C terminal modification.
  • the cationic domain at the C terminus comprises a C-terminal modification.
  • the cationic domain at the N terminus comprises a N-terminal modification.
  • the cationic domain at the C terminus comprises a linker group, suitably, the cationic domain at the C terminus comprises a C-terminal beta-alanine.
  • the cationic domain at the N terminus is N-acetylated.
  • the peptide of the present invention is defined as having a total length of 40 amino acid residues or less.
  • the peptide may therefore be regarded as an oligopeptide.
  • the peptide has a total length of between 3-30 amino acid residues, suitably of between 5-25 amino acid residues, of between 10-25 amino acid residues, of between 13-23 amino acid residues, of between 15-20 amino acid residues.
  • the peptide has a total length of at least 12, at least 13, at least 14, at least 15, at least 16, at least 17 amino acid residues.
  • the peptide is capable of penetrating cells.
  • the peptide may therefore be regarded as a cell-penetrating peptide.
  • the peptide is for attachment to a therapeutic molecule.
  • the peptide is for transporting a therapeutic molecule into a target cell.
  • the peptide is for delivering a therapeutic molecule into a target cell.
  • the peptide may therefore be regarded as a carrier peptide.
  • the peptide is capable of penetrating into cells and tissues, suitably into the nucleus of cells. Suitably into muscle tissues.
  • the peptide may be selected from any of the following sequences:
  • HBRRBRFQILYRBHBH (SEQ ID NO:47)
  • the peptide may be selected from any of the following additional sequences:
  • RBRRBRWPWWBRBR (SEQ ID NQ:60)
  • RBRRBRWWPWBRBR (SEQ ID NO:61)
  • the peptide consists of one of the following sequences:
  • the peptide consists of the following sequence: RBRRBRFQILYBRBR (SEQ ID NO:35).
  • the peptide consists of the following sequence: RBRRBRRFQILYRBHBH (SEQ ID NO:37).
  • the peptide consists of the following sequence: RBRRBRFQILYRBHBH (SEQ ID NO:44).
  • the peptide of the invention may be covalently linked to a therapeutic molecule in order to provide a conjugate.
  • the therapeutic molecule may be any molecule for treatment of a disease.
  • the therapeutic molecule may be selected from: a nucleic acid, peptide nucleic acid, antisense oligonucleotide (such as PNA, PMO), mRNA, gRNA (for example in the use of CRISPR/Cas9 technology), short interfering RNA, micro RNA, antagomiRNA, peptide, cyclic peptide, protein, pharmaceutical, drug, or nanoparticle.
  • the therapeutic molecule is an antisense oligonucleotide.
  • the antisense oligonucleotide is comprised of a phosphorodiamidate morpholino oligonucleotide (PMO).
  • PMO phosphorodiamidate morpholino oligonucleotide
  • the oligonucleotide may be a modified PMO or any other charge-neutral oligonucleotide such as a peptide nucleic acid (PNA), a chemically modified PNA such as a gamma-PNA (Bahai, Nat.Comm. 2016), oligonucleotide phosphoramidate (where the non bridging oxygen of the phosphate is substituted by an amine or alkylamine such as those described in WO2016028187A1 , or any other partially or fully charge-neutralized oligonucleotide.
  • PNA peptide nucleic acid
  • gamma-PNA gamma-PNA
  • oligonucleotide phosphoramidate where the non bridging oxygen of the phosphate is substituted by an amine or alkylamine such as those described in WO2016028187A1 , or any other partially or fully charge-neutralized oligonucleotide.
  • the therapeutic antisense oligonucleotide sequence may be selected from any that are available, for example antisense oligonucleotides for exon skipping in DMD are described in https://research-repository.uwa.edu.au/en/publications/antisense-oligonucleotide-induced- exon-skipping-across-the-human- , or a therapeutic antisense oligonucleotide complementary to the ISSN1 or IN7 sequence for the treatment of SMA are described in Zhou, HGT, 2013; and Hammond et al, 2016; and Osman et al, HMG, 2014.
  • the antisense oligonucleotide sequence is for inducing exon skipping for use in the treatment of DMD. .
  • the antisense oligonucleotide sequence is for inducing exon skipping in the dystrophin gene for use in the treatment of DMD.
  • the antisense oligonucleotide sequence may induce exon skipping of one or multiple exons.
  • the antisense oligonucleotide sequence is for inducing exon skipping of a single exon of the dystrophin gene for use in the treatment of DMD.
  • the single exon is selected from any exon implicated in DMD, which may be any exon in the dystrophin gene, such as for example, exon 45, 51 or 53.
  • PMO oligonucleotides of any sequence may be purchased (for example from Gene Tools Inc, USA).
  • the therapeutic molecule of the conjugate is an oligonucleotide complementary to the pre-mRNA of a gene target.
  • the oligonucleotide complementary to the pre-mRNA of a gene target gives rise to a steric blocking event that alters the pre-mRNA leading to an altered mRNA and hence a protein of altered sequence.
  • the gene target is the dystrophin gene.
  • the steric blocking event may be exon inclusion or exon skipping.
  • the steric blocking event is exon skipping, suitably exon skipping of a single exon of the dystrophin gene.
  • lysine residues may be added to one or both ends of a therapeutic molecule (such as a PMO or PNA) before attachment to the peptide to improve water solubility.
  • the therapeutic molecule has a molecular weight of less than 5,000 Da, suitably less than 3,000 Da or suitably less than 1 ,000 Da.
  • the peptide is covalently linked to the therapeutic molecule at the C-terminus.
  • the peptide is covalently linked to the therapeutic molecule through a linker if required.
  • the linker may act as a spacer to separate the peptide sequence from the therapeutic molecule.
  • the linker may be selected from any suitable sequence.
  • the linker is present between the peptide and the therapeutic molecule.
  • the linker is a separate group to the peptide and the therapeutic molecule. Accordingly, the linker may comprise artificial amino acids.
  • the conjugate comprises the peptide covalently linked via a linker to a therapeutic molecule. In one embodiment, the conjugate comprises the following structure:
  • the conjugate consists of the following structure:
  • the conjugate comprises a peptide selected from one of the following sequences: RBRRBRFQILYBRBR (SEQ ID NO:35), RBRRBRRFQILYRBHBH (SEQ ID NO:37) and RBRRBRFQILYRBHBH (SEQ ID NO:44).
  • the peptide may further comprise N-terminal modifications as described above.
  • Suitable linkers include, for example, a C-terminal cysteine residue that permits formation of a disulphide, thioether or thiol-maleimide linkage, a C-terminal aldehyde to form an oxime, a click reaction or formation of a morpholino linkage with a basic amino acid on the peptide or a carboxylic acid moiety on the peptide covalently conjugated to an amino group to form a carboxamide linkage.
  • the linker is between 1- 5 amino acids in length.
  • the linker may comprise any linker that is known in the art.
  • the linker is selected from any of the following sequences: G, BC, XC, C, GGC, BBC, BXC, XBC, X, XX, B, BB, BX and XB.
  • X is 6-aminohexanoic acid.
  • the linker may be a polymer, such as for example PEG.
  • the linker is beta-alanine.
  • the peptide is conjugated to the therapeutic molecule through a carboxamide linkage.
  • the linker of the conjugate may form part of the therapeutic molecule to which the peptide is attached.
  • the attachment of the therapeutic molecule may be directly linked to the C-terminus of the peptide.
  • no linker is required.
  • the peptide may be chemically conjugated to the therapeutic molecule.
  • Chemical linkage may be via a disulphide, alkenyl, alkynyl, aryl, ether, thioether, triazole, amide, carboxamide, urea, thiourea, semicarbazide, carbazide, hydrazine, oxime, phosphate, phosphoramidate, thiophosphate, boranophosphate, iminophosphates, or thiol-maleimide linkage, for example.
  • cysteine may be added at the N- terminus of a therapeutic molecule to allow for disulphide bond formation to the peptide, or the N-terminus may undergo bromoacetylation for thioether conjugation to the peptide.
  • the peptide of the invention may equally be covalently linked to an imaging molecule in order to provide a conjugate.
  • the imaging molecule may be any molecule that enables visualisation of the conjugate.
  • the imaging molecule may indicate the location of the conjugate.
  • the location of the conjugate in vitro or in vivo.
  • a method of monitoring the location of a conjugate comprising an imaging molecule comprising: administering the conjugate to a subject and imaging the subject to locate the conjugate.
  • imaging molecules include detection molecules, contrast molecules, or enhancing molecules.
  • Suitable imaging molecules may be selected from radionuclides; fluorophores; nanoparticles (such as a nanoshell); nanocages; chromogenic agents (for example an enzyme), radioisotopes, dyes, radiopaque materials, fluorescent compounds, and combinations thereof.
  • imaging molecules are visualised using imaging techniques, these may be cellular imaging techniques or medical imaging techniques.
  • Suitable cellular imaging techniques include image cytometry, fluorescent microscopy, phase contrast microscopy, SEM, TEM, for example.
  • Suitable medical imaging techniques include X-ray, fluoroscopy, MRI, scintigraphy, SPECT, PET, CT, CAT, FNRI, for example.
  • the imaging molecule may be regarded as a diagnostic molecule.
  • a diagnostic molecule enables the diagnosis of a disease using the conjugate.
  • diagnosis of a disease may be achieved through determining the location of the conjugate using an imaging molecule.
  • a method of diagnosis of a disease comprising administering an effective amount of a conjugate comprising an imaging molecule to a subject and monitoring the location of the conjugate.
  • the peptide of the invention may be covalently linked to a therapeutic molecule and an imaging molecule in order to provide a conjugate.
  • the conjugate is capable of penetrating into cells and tissues, suitably into the nucleus of cells. Suitably into muscle tissues.
  • the conjugate of the invention may formulated into a pharmaceutical composition.
  • the pharmaceutical composition comprises a conjugate of the invention.
  • the pharmaceutical composition may further comprise a pharmaceutically acceptable diluent, adjuvant or carrier.
  • Suitable pharmaceutically acceptable diluents, adjuvants and carriers are well known in the art.
  • the phrase "pharmaceutically acceptable” refers to those ligands, materials, formulations, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier refers to a pharmaceutically acceptable material, formulation or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the conjugate from one organ or portion of the body, to another organ or portion of the body.
  • a pharmaceutically acceptable material, formulation or vehicle such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the conjugate from one organ or portion of the body, to another organ or portion of the body.
  • Each cell-penetrating peptide must be “acceptable” in the sense of being compatible with the other components of the composition e.g. the peptide and therapeutic molecule, and not injurious to the individual.
  • Lyophilized compositions which may be reconstituted and administered, are also within the scope of the present composition.
  • compositions may be, for example, excipients, vehicles, diluents, and combinations thereof.
  • the compositions may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections, drop infusion preparations, or suppositories.
  • the active compound i.e. conjugate
  • the active compound may be mixed with any conventional additive, such as an excipient, a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, a coating agent, or combinations thereof.
  • compositions of the present disclosure can further include additional known therapeutic agents, drugs, modifications of compounds into prodrugs, and the like for alleviating, mediating, preventing, and treating the diseases, disorders, and conditions described herein under medical use.
  • the pharmaceutical composition is for use as a medicament.
  • a pharmaceutical composition according to the fourth aspect for use as a medicament.
  • a method of treating a subject for a disease condition comprising administering an effective amount of a pharmaceutical composition according to the fourth aspect to the subject.
  • the conjugate comprising the peptide of the invention may be used as a medicament for the treatment of a disease.
  • the medicament may be in the form of a pharmaceutical composition as defined above.
  • a method of treatment of a patient or subject in need of treatment for a disease condition comprising the step of administering a therapeutically effective amount of the conjugate to the patient or subject.
  • the medical treatment requires delivery of the therapeutic molecule into a cell, suitably into the nucleus of the cell.
  • Diseases to be treated may include any disease where improved penetration of the cell and/or nuclear membrane by a therapeutic molecule may lead to an improved therapeutic effect.
  • the conjugate is for use in the treatment of diseases of the neuromuscular system.
  • Conjugates comprising peptides of the invention are suitable for the treatment of genetic diseases of the neuromuscular system. Conjugates comprising peptides of the invention are suitable for the treatment of genetic neuromuscular diseases.
  • a conjugate according to the second aspect for use in the treatment of genetic diseases of the neuromuscular system.
  • the conjugate is for use in the treatment of hereditary genetic diseases.
  • the conjugate is for use in the treatment of hereditary genetic diseases of the neuromuscular system.
  • the conjugate is for use in the treatment of hereditary genetic neuromuscular diseases.
  • the conjugate is for use in the treatment of hereditary X-linked genetic diseases of the neuromuscular system.
  • the conjugate is for use in the treatment of hereditary X-linked neuromuscular diseases.
  • the conjugate is for use in the treatment of diseases caused by splicing deficiencies.
  • the therapeutic molecule may comprise an oligonucleotide capable of preventing or correcting the splicing defect and/or increasing the production of correctly spliced mRNA molecules.
  • the conjugate is for use in the treatment of any of the following diseases: Duchenne Muscular Dystrophy (DMD), Bucher Muscular Dystrophy (BMD), Menkes disease, Beta- thalassemia, dementia, Parkinson’s Disease, Spinal Muscular Atrophy (SMA), myotonic dystrophy (DM), Huntington’s Disease, Hutchinson-Gilford Progeria Syndrome, Ataxia- telangiectasia, or cancer.
  • DMD Duchenne Muscular Dystrophy
  • BMD Bucher Muscular Dystrophy
  • Menkes disease Beta- thalassemia
  • dementia dementia
  • Parkinson’s Disease Spinal Muscular Atrophy
  • SMA Spinal Muscular Atrophy
  • DM myotonic dystrophy
  • Huntington’s Disease Hutchinson-Gilford Progeria Syndrome
  • Ataxia- telangiectasia or cancer.
  • the conjugate is for use in the treatment of DMD.
  • a conjugate according to the second aspect for use in the treatment of DMD is provided.
  • the therapeutic molecule of the conjugate is operable to increase expression of the dystrophin protein.
  • the therapeutic molecule of the conjugate is operable to increase the expression of functional dystrophin protein.
  • the conjugate increases dystrophin expression by 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%.
  • the conjugate increases dystrophin expression by up to 50%.
  • the conjugate restores dystrophin protein expression by 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%.
  • the conjugate restores dystrophin protein expression by up to 50%.
  • the conjugate restores dystrophin protein function by 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%.
  • the conjugate restores dystrophin protein function by up to 50%.
  • the therapeutic molecule of the conjugate is operable to do so by causing skipping of one or more exons during dystrophin transcription.
  • the therapeutic molecule of the conjugate causes 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85% skipping of one or more exons of the dystrophin gene.
  • the therapeutic molecule of the conjugate causes up to 50% skipping of one or more exons of the dystrophin gene.
  • the patient or subject to be treated may be any animal or human.
  • the patient or subject may be a non-human mammal.
  • the patient or subject may be male or female. In one embodiment, the subject is male.
  • the patient or subject to be treated may be any age.
  • the patient or subject to be treated is aged between 0-40 years, suitably 0-30, suitably 0-25, suitably 0-20 years of age.
  • the conjugate is for administration to a subject systemically for example by intramedullary, intrathecal, intraventricular, intravitreal, enteral, parenteral, intravenous, intra arterial, intramuscular, intratumoral, subcutaneous oral or nasal routes.
  • the conjugate is for administration to a subject intravenously.
  • the conjugate is for administration to a subject intravenously by injection.
  • the conjugate is for administration to a subject in a "therapeutically effective amount", by which it is meant that the amount is sufficient to show benefit to the individual.
  • a therapeuticically effective amount by which it is meant that the amount is sufficient to show benefit to the individual.
  • the actual amount administered, and rate and time-course of administration, will depend on the nature and severity of the disease being treated. Decisions on dosage are within the responsibility of general practitioners and other medical doctors. Examples of the techniques and protocols can be found in Remington's Pharmaceutical Sciences, 20th Edition, 2000, pub. Lippincott, Williams & Wlkins.
  • Exemplary doses may be between 0.01 mg/kg and 50mg/kg, 0.05mg/kg and 40mg/kg, 0.1 mg/kg and 30mg/kg, 0.5mg/kg and 18mg/kg, 1 mg/kg and 16mg/kg, 2mg/kg and 15mg/kg, 5mg/kg and 10mg/kg, 10mg/kg and 20mg/kg, 12mg/kg and 18mg/kg, 13mg/kg and 17mg/kg.
  • the dosage of the conjugates of the present invention is an order or magnitude lower than the dosage required to see any effect from the therapeutic molecule alone.
  • one or more markers of toxicity are significantly reduced compared to prior conjugates using currently available peptide carriers
  • Suitable markers of toxicity may be markers of nephrotoxicity.
  • Suitable markers of toxicity include KIM-1 , NGAL, BUN, creatinine, alkaline phosphatase, alanine transferase, and aspartate aminotransferase.
  • the level of at least one of KIM-1 , NGAL, and BUN is reduced after administration of the conjugates of the present invention when compared to prior conjugates using currently available peptide carriers.
  • the levels of each of KIM-1 , NGAL, and BUN are reduced after administration of the conjugates of the present invention when compared to prior conjugates using currently available peptide carriers.
  • the levels of the or each marker/s is significantly reduced when compared to prior conjugates using currently available peptide carriers.
  • the levels of the or each marker/s is reduced by up to 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50% after administration of the conjugates of the present invention when compared to prior conjugates using currently available peptide carriers.
  • the toxicity of the peptides and therefore the resulting conjugates is significantly reduced compared to prior cell-penetrating peptides and conjugates.
  • KIM-1 and NGAL-1 are markers of toxicity and these are significantly reduced by up to 120 times compared to prior conjugates using currently available peptide carriers.
  • Peptides of the invention may be produced by any standard protein synthesis method, for example chemical synthesis, semi-chemical synthesis or through the use of expression systems. Accordingly, the present invention also relates to the nucleotide sequences comprising or consisting of the DNA coding for the peptides, expression systems e.g. vectors comprising said sequences accompanied by the necessary sequences for expression and control of expression, and host cells and host organisms transformed by said expression systems.
  • nucleic acid encoding a peptide according to the present invention is also provided.
  • the nucleic acids may be provided in isolated or purified form.
  • An expression vector comprising a nucleic acid encoding a peptide according to the present invention is also provided.
  • the vector is a plasmid.
  • the vector comprises a regulatory sequence, e.g. promoter, operably linked to a nucleic acid encoding a peptide according to the present invention.
  • the expression vector is capable of expressing the peptide when transfected into a suitable cell, e.g. mammalian, bacterial or fungal cell.
  • a host cell comprising the expression vector of the invention is also provided.
  • Expression vectors may be selected depending on the host cell into which the nucleic acids of the invention may be inserted. Such transformation of the host cell involves conventional techniques such as those taught in Sambrook et al [Sambrook, J., Russell, D. (2001) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY, USA] Selection of suitable vectors is within the skills of the person knowledgeable in the field. Suitable vectors include plasmids, bacteriophages, cosmids, and viruses.
  • the peptides produced may be isolated and purified from the host cell by any suitable method e.g. precipitation or chromatographic separation e.g. affinity chromatography.
  • Suitable vectors, hosts and recombinant techniques are well known in the art.
  • operably linked may include the situation where a selected nucleotide sequence and regulatory nucleotide sequence are covalently linked in such a way as to place the expression of a nucleotide coding sequence under the control of the regulatory sequence, as such, the regulatory sequence is capable of effecting transcription of a nucleotide coding sequence which forms part or all of the selected nucleotide sequence.
  • the resulting transcript may then be translated into a desired peptide.
  • Figure 1 shows the in vitro exon 23 skipping efficacy of some of the DPEP1 series of peptides conjugated to an antisense therapeutic PMO at 0.25mM, O.dmM and 1 mM in H2K-mdx cells as measured by densitometry analysis of nested RT-PCR (Error bars: standard deviation, n33);
  • Figure 2 shows the in vitro exon 23 skipping efficacy of some of the DPEP3 series of peptides conjugated to an antisense therapeutic PMO at 0.25mM, O.dmM and 1 mM in H2K-mdx cells as measured by densitometry analysis of nested RT-PCR (Error bars: standard deviation, n33);
  • Figure 10 shows the in vivo efficacy of exon 23 skipping assessed by qRT-PCR in (A) tibalis anterior, (B) diaphragm and (C) heart of C57BL/6 mice following a single 30 mg/kg intravenous administration of various DPEP peptides conjugated to an antisense therapeutic PMO, in comparison with currently available peptide carriers conjugated to the same antisense therapeutic PMO and saline.
  • qPCR analysis of exon 23 exclusion was assessed in (A) tibialis anterior, (B) diaphragm and (C) heart at 7 days post-administration.
  • Figure 19 shows the (A) alanine transferase (ALT), (B) alkaline phosphatase (ALP) and (C) aspartate aminotransferase (AST) levels assessed in serum from C57BL6 female mice, who were administered by bolus IV (tail vein) injection of different DPEP1/3-[CAG] 7 PMO conjugates, at day 7 post-injection collection compared to saline.
  • ALP, ALT, AST levels were similar to saline control injections in comparison to the fold increases induced by the prior Pip series of peptide carriers.
  • HPLC grade acetonitrile, methanol and synthesis grade N-methyl-2-pyrrolidone (NMP) were purchased from Fisher Scientific (Loughborough, UK).
  • Peptide synthesis grade A/,/ ⁇ /-dimethylformamide (DMF) and diethyl ether were obtained from VWR (Leicestershire, UK).
  • Piperidine and trifluoroacetic acid (TFA) were obtained from Alfa Aesar (Heysham, England).
  • PMO was purchased from Gene Tools Inc. (Philomath, USA).
  • Chicken Embryo Extract and horse serum were obtained from Sera Laboratories International Ltd (West Wales, UK). Interferon was obtained from Roche Applied Science (Penzberg, Germany).
  • Peptides were either prepared on a 10 pmol scale using an Intavis Parallel Peptide Synthesizer or on a 100 pmol scale using a CEM Liberty BlueTM Peptide Synthesizer (Buckingham, UK) using Fmoc ⁇ -Ala-OH preloaded Wang resin (0.19 or 0.46 mmol g _1 , Merck Millipore) by applying standard Fmoc chemistry and following manufacturer’s recommendations.
  • double coupling steps were used with a PyBOP/NMM coupling mixture followed by acetic anhydride capping after each step.
  • the peptide resin was washed with DMF (3 x 20 mL) and DCM (3 x 20 mL).
  • the peptides were cleaved from the solid support by treatment with a cleavage cocktail consisting of trifluoroacetic acid (TFA): H2O: triisopropylsilane (TIPS) (95%: 2.5%: 2.5%: 3-10 mL) for 3 h at room temperature.
  • TFA trifluoroacetic acid
  • H2O triisopropylsilane
  • TFA triisopropylsilane
  • the crude peptide pellet was washed thrice with cold diethyl ether (3 x 15 mL) and purified by RP-HPLC using a Varian 940-LC HPLC System fitted with a 445-LC Scale-up module and 440-LC fraction collector.
  • Peptides were purified by semi-preparative HPLC on an RP-C18 column (10 x 250 mm, Phenomenex Jupiter) using a linear gradient of CH3CN in 0.1 % TFA/H2O with a flow rate of 15 mL min 1 . Detection was performed at 220 nm and 260 nm. The fractions containing the desired peptide were combined and lyophilized to yield the peptide as a white solid.
  • Table 1 peptides as synthesised for testing in the examples with N-terminal acetylation and C-terminal beta-alanine linker.
  • Pip9b2 and R6Gly are comparative peptides.
  • R6Gly uses a C- terminal glycine as a linker.
  • a 25-mer PMO antisense sequence for mouse dystrophin exon-23 (GGCCAAACCTCGGCTTACCTGAAAT (SEQ ID NO:90)) was used.
  • the peptide was conjugated to the 3’-end of the PMO through its C-terminal carboxyl group. This was achieved using 2.3 and 2 equivalents of PyBOP and HOAt in NMP respectively in the presence of 2.3 equivalents of DIPEA over peptide and 2.5 fold excess of peptide over PMO dissolved in DMSO.
  • 2.3 equivalents of HBTU were used in place of PyBOP for activation of the C-terminal carboxyl group of the peptide.
  • the PMO-peptide conjugates were purified on an ion exchange column (Resource S 4 ml_, GE Healthcare) using a linear gradient of sodium chloride (0 to 1 M) in sodium phosphate buffer (25 mM, pH 7.0) containing 20 % CH3CN at a flow rate of 4 ml_ min-1.
  • the fractions containing the desired compound were combined and lyophilized to yield the peptide-PMO derivative as a white solid.
  • the removal of excess salts from the peptide-PMO conjugate was afforded through the filtration of the fractions collected after ion exchange using an Amicon® ultra-15 3K centrifugal filter device.
  • the conjugate was lyophilized and analyzed by MALDI-TOF.
  • the conjugates were dissolved in sterile water and filtered through a 0.22 pm cellulose acetate membrane before use.
  • concentration of peptide-PMO was determined by the molar absorption of the conjugates at 265 nm in 0.1 N HCI solution (see Table 2 for yields).
  • Murine H2k mdx myoblasts were cultured in gelatin (0.01 %) coated flasks at 33 °C, under 10% CO2 in Dulbecco’s modified Eagles medium (DM EM PAA laboratories) supplemented with 20% heat-inactivated fetal bovine serum (FBS Gold, PAA laboratories), 2% chicken embryo extract (Seralab), 1 % penicillin-streptomycin-neomycin antibiotic mixture (PSN, Gibco) and 3pg/pL g-interferon (Roche). Cells were seeded in gelatin (0.01 %) coated 24-well plates at a density of 2 x 10 5 cell/mL and left for 2 days at 33 °C, 10% CO2. To differentiate into myotubes, cells were further grown in DM EM supplemented with 5% horse serum (Sigma) and 1 % PSN at 37 °C, under 5% C0 2 for 2 days.
  • DM EM PAA laboratories Dulbecco’s modified Eagles medium (
  • RNA Total cellular RNA was extracted using TRI reagent with an extra further precipitation with ethanol.
  • the RNA (400 ng) was used as a template for RT-PCR using a OneStep RT-PCR Kit (Roche, Indianapolis, USA).
  • primer sequences refer to Table 4.
  • the cycle conditions for the initial reverse transcription were 50 °C for 30 min and 94 °C for 7 min for 1 cycle followed by 30 cycles of 94 °C for 20 sec, 55 °C for 40 sec and 68 °C for 80 sec.
  • One microlitre of the RT- PCR product was used as template for the second PCR step.
  • the amplification was carried out using 0.5 U of SuperTAQ in 25 cycles at 94 °C for 30 sec, 55 °C for 1 min and 72 °C for 1 min. the products were separated by electrophoresis using 1.5% agarose gel.
  • the images of agarose gels were taken on a Molecular Imager ChemiDocTMXRS + imaging system (BioRad, UK) and the images were analysed using Image Lab (V4.1).
  • Microsoft Excel was used to analyse and plot the exon-skipping assay data, which were expressed as the percentage of exon-23 skipping from at least three independent experiments.
  • Peptides were synthesized on a 100 pmol scale using a CEM Liberty BlueTM microwave Peptide Synthesizer (Buckingham, UK) and Fmoc chemistry following manufacturer’s recommendations.
  • the side chain protecting groups used were labile to trifluoroacetic acid treatment and the peptide was synthesized using a 5-fold excess of Fmoc-protected amino acids (0.25 mmol) that were activated using PyBOP (5-fold excess) in the presence of DIPEA.
  • Piperidine (20 % v/v in DMF) was used to remove N-Fmoc protecting groups. The coupling was carried out once at 75 °C for 5 min at 60-watt microwave power except for arginine residues, which were coupled twice each.
  • each deprotection reaction was carried out at 75 °C twice, once for 30 sec and then once for 3 min at 35-watt microwave power.
  • the resin was washed with DMF (3 x 50 mL) and the N-terminus of the solid phase bound peptide was acetylated with acetic anhydride in the presence of DI PEA at room temperature. After acetylation of the N-terminus, the peptide resin was washed with DMF (3 x 20 mL) and DCM (3 x 20 mL).
  • the peptide was cleaved from the solid support by treatment with a cleavage cocktail consisting of trifluoroacetic acid (TFA): H 2 O: triisopropylsilane (TIPS) (95%: 2.5%: 2.5%, 10 mL) for 3 h at room temperature. Excess TFA was removed by sparging with nitrogen. The cleaved peptide was precipitated via the addition of ice-cold diethyl ether and centrifuged at 3000 rpm for 5 min.
  • TFA trifluoroacetic acid
  • H 2 O triisopropylsilane
  • the crude peptide pellet was washed thrice with cold diethyl ether (3 c 40 mL) and purified by RP-HPLC using a Varian 940-LC HPLC System fitted with a 445-LC Scale-up module and 440-LC fraction collector.
  • Peptides were purified by semi preparative HPLC on an RP-C18 column (10 x 250 mm, Phenomenex Jupiter) using a linear gradient of CH 3 CN in 0.1 % TFA/H 2 O with a flow rate of 15 mL min 1 . Detection was performed at 220 nm and 260 nm.
  • NMP N- methylpyrrolidone
  • PyBOP 76.6 mI_ of 0.3 M in NMP
  • HOAt 66.7 mI_ of 0.3 M NMP
  • DIPEA 4.0 mI_
  • PMO 400 mI_ of 10 mM in DMSO
  • the reaction was purified on a cation exchange chromatography column (Resource S 6 mL column, GE Healthcare) using a linear gradient of sodium chloride (0 to 1 M) in sodium phosphate buffer (25 mM, pH 7.0) containing 20 % CH3CN at a flow rate of 6 ml_ min-1.
  • the removal of excess salts from the peptide-PMO conjugate was afforded through the filtration of the fractions collected after ion exchange using an Amicon® ultra-15 3K centrifugal filter device.
  • the conjugate was lyophilized and analyzed by MALDI-TOF.
  • the conjugates were dissolved in sterile water and filtered through a 0.22 pm cellulose acetate membrane before use.
  • the concentration of peptide-PMO was determined by the molar absorption of the conjugates at 265 nm in 0.1 N HCI solution. Overall yields (Table 3) were 25-36% based on PMO.
  • mice were housed in a minimal disease facility; the environment was temperature controlled with a 12 hour light-dark cycle. All animals received commercial rodent chow and water ad libitum.
  • mice were restrained prior to a single tail vein injection of 10mg/kg of P-PMO.
  • P-PMO 10mg/kg of P-PMO.
  • mice were sacrificed and TA, heart and diaphragm muscles removed and snap frozen in dry-ice cooled isopentane and stored at -80 °C.
  • one-third of the muscle (for TA and diaphragm) or ninety 7 pm thick transverse cryosections (for heart) were lysed in 300 mI buffer (50mM Tris pH 8, 150mM NaCI, 1 %NP40, 0.5% sodium deoxycholate, 10% SDS and protease/phosphatase inhibitors) prior to centrifuging at 13000 rpm (Heraeus, #3325B) for 10 min. Supernatant was collected and heated at 100°C for 3 min. Protein was quantified by BCA method and 40 pg protein/sample were resolved in a NuPage a 3-8% Tris-Acetate gel as previously described (19).
  • Proteins were transferred to a 0.45 pm pore size PVDF membrane for 1 h at 30V followed by 1 h at 100V, and probed with monoclonal anti-dystrophin (1 :200, NCL-DYS1 , Novocastra) and anti-vinculin (loading control, 1 :100 000, hVIN-1 , Sigma) antibodies as previously described (37).
  • monoclonal anti-dystrophin (1 :200, NCL-DYS1 , Novocastra
  • anti-vinculin loading control, 1 :100 000, hVIN-1 , Sigma
  • Secondary antibody IRDye 800CW goat anti-mouse was used at a dilution of 1 :20000 (LiCOR).
  • the levels of dystrophin restoration in P-PMO treated mdx mice were expressed as relative to the levels of C57BL/10 wildtype control mice, considered as 100%.
  • a standard curve was generated by including 5 serial C57BL/10 protein dilutions in parallel to the P-PMO treated mdx samples. Dilution series were as follows: 75%, 40%, 15%, 5% or 0% respectively of the 40 pg total protein loaded per lane were from C57BL/10 protein lysates and the remaining from un-treated mdx protein lysates. These standards were aliquoted and used in each western blot in parallel to the treated mdx samples.
  • Dystrophin intensity quantification was performed by Fluorescence Odyssey imaging system and normalized by calculating the ratio to the Vinculin fluorescence intensity in all samples. Standard normalized values were plotted against their known concentration of dystrophin to obtain the mathematical expression of best fit and this expression used to interpolate the normalized values of each sample of P-PMO treated mdx mice.
  • Primer/probes were synthesised by Integrated DNA Technologies and designed to amplify a region spanning exon 23 - 24 representing unskipped product (mDMD23-24, see Table 4), or to amplify specifically transcripts lacking exon 23 using a probe spanning the boundary of exon 22 and 24 (mDMD22-24).
  • Levels of respective transcripts were determined by calibration to standard curves prepared using known transcript quantities, and skipping percentages derived by [skip]/[skip+unskip].
  • Table 4 Primer and probe sequences for quantification of exon 23 skipping by nested RT-PCR or quantitative RT-PCR methods.
  • mice Female C57BL/6 mice aged 8-10 weeks were administered a single 30 mg/kg dose of peptide- PMO in 0.9% saline by bolus intravenous tail vein injection.
  • Urine was non-invasively collected under chilled conditions at Day 2 and Day 7 post-administration following 20 hours housing in metabolic cages (Tecniplast, UK). Serum was collected from jugular vein at Day 7 at necropsy, as was tibialis anterior, diaphragm and heart tissue.
  • Urinary levels of KIM-1 (Kidney injury molecule-1) and NGAL (Neutrophil Gelatinase- Associated Lipocalin) were quantified by ELISA (KIM-1 R&D cat# MKM100, NGAL R&D cat# MLCN20) following appropriate dilution of urine to fit standard curves. Values were normalised to urinary creatinine levels that were quantified at MRC Harwell Institute, Mary Lyon Centre, Oxfordshire, UK. Serum blood urea nitrogen levels were quantified at MRC Harwell Institute, Mary Lyon Centre, Oxfordshire, UK.
  • exon skipping activity remains high for all of the DPEP peptide conjugates in TA and diaphragm (figure 10 and 12) at higher doses of 30 and 50mg/kg, which when corroborated with the reduced levels of kidney damage markers, suggests a wider therapeutic index for these compounds because toxicity markers are many-fold lower. It is also notable that all of the DPEP peptide conjugates have higher activity than the known R6Gly comparator in a conjugate, whilst at least maintaining similar levels of toxicity markers, and similar activity to the known PIP peptide comparator in a conjugate whilst having much lower levels of toxicity markers. In some cases, the DPEP peptide conjugates of the invention display not only increased activity compared to the known R6Gly conjugate but also reduced toxicity markers.
  • the DPEP1 and 3 peptides of the invention provide promising cell-penetrating peptides for improving the efficacy and reducing the toxicity of therapeutic conjugates for the treatment of neuromuscular disorders in humans.
  • HPLC grade acetonitrile, methanol and synthesis grade N-methyl-2-pyrrolidone (NMP) were purchased from Fisher Scientific (Loughborough, UK).
  • Peptide synthesis grade N,N-dimethylformamide (DMF) and diethyl ether were obtained from VWR (Leicestershire, UK).
  • Piperidine and trifluoroacetic acid (TFA) were obtained from Alfa Aesar (Heysham, England).
  • PMO was purchased from Gene Tools Inc. (Philomath, USA). All other reagents were obtained from Sigma-Aldrich (St. Louis, MO, USA) unless otherwise stated.
  • MALDI-TOF mass spectrometry was carried out using a Voyager DE Pro BioSpectrometry workstation. A stock solution of 10 mg mL-1 of a-cyano-4-hydroxycinnamic acid or sinapinic acid in 50% acetonitrile in water was used as matrix. Error bars are ⁇ 0.1 %
  • Peptides were either prepared on a 10 pmol scale using an Intavis Parallel Peptide Synthesizer or on a 100 pmol scale using a CEM Liberty BlueTM Peptide Synthesizer (Buckingham, UK) using Fmoc ⁇ -Ala-OH preloaded Wang resin (0.19 or 0.46 mmol g-1 , Merck Millipore) by applying standard Fmoc chemistry and following manufacturer’s recommendations.
  • double coupling steps were used with a PyBOP/NMM coupling mixture followed by acetic anhydride capping after each step.
  • the peptide resin was washed with DMF (3 x 20 mL) and DCM (3 x 20 mL).
  • the peptides were cleaved from the solid support by treatment with a cleavage cocktail consisting of trifluoroacetic acid (TFA): H20: triisopropylsilane (TIPS) (95%: 2.5%: 2.5%: 3-10 mL) for 3 h at room temperature.
  • TFA trifluoroacetic acid
  • H20 triisopropylsilane
  • TFA triisopropylsilane
  • the crude peptide pellet was washed thrice with cold diethyl ether (3 x 15 mL) and purified by RP-HPLC using a Varian 940-LC HPLC System fitted with a 445-LC Scale-up module and 440-LC fraction collector.
  • Peptides were purified by semi-preparative HPLC on an RP-C18 column (10 x 250 mm, Phenomenex Jupiter) using a linear gradient of CH3CN in 0.1 % TFA/H20 with a flow rate of 15 mL min-1. Detection was performed at 220 nm and 260 nm. The fractions containing the desired peptide were combined and lyophilized to yield the peptide as a white solid (see Table 5 for yields).
  • a 21-mer PMO antisense sequence for triplet repeat sequences (CAGCAGCAGCAGCAGCAGCAG (SEQ ID NO.107) otherwise known as [CAG]7 was used.
  • the peptide was conjugated to the 3’-end of the PMO through its C-terminal carboxyl group. This was achieved using 2.5 and 2 equivalents of PyBOP and HOAt in NMP respectively in the presence of 2.5 equivalents of DIPEA and 2.5 fold excess of peptide over PMO dissolved in DMSO was used.
  • the PMO-peptide conjugates were purified on an ion exchange column (Resource S 4 ml_, GE Healthcare) using a linear gradient of sodium phosphate buffer (25 mM, pH 7.0) containing 20 % CH3CN. A sodium chloride solution (1 M) was used to elute the conjugate from the column at a flow rate of either 4 ml_ min-1 or 6ml_ min-1. The fractions containing the desired compound were combined desalted immediately. The removal of excess salts from the peptide-PMO conjugate was afforded through the filtration of the fractions collected after ion exchange using an Amicon® ultra-15 3K centrifugal filter device. The conjugate was lyophilized and analyzed by MALDI-TOF.
  • the conjugates were dissolved in sterile water and filtered through a 0.22 pm cellulose acetate membrane before use.
  • concentration of peptide-PMO was determined by the molar absorption of the conjugates at 265 nm in 0.1 N HCI solution (see Table 6 for yields).
  • Peptides were synthesized and conjugated to PMO as described previously.
  • the PMO sequence targeting CUG/CTG expanded repeats (5 -CAGCAGCAGCAGCAGCAGCAGCAGCAG-3' (SEQ ID NO: 107)) was purchased from Gene Tools LLC. This is a [CAG]7 PMO as referenced elsewhere herein.
  • Immortalized myoblasts from healthy individual or DM1 patient with 2600 CTG repeats were cultivated in a growth medium consisting of a mix of M 199: DM EM (1 :4 ratio; Life technologies) supplemented with 20% FBS (Life technologies), 50 pg/ml gentamycin (Life technologies), 25 pg/ml fetuin, 0.5 ng/ml bFGF, 5 ng/ml EGF and 0.2 pg/ml dexamethasone (Sigma-Aldrich). Myogenic differentiation was induced by switching confluent cell cultures to DMEM medium supplemented with 5 pg/ml insulin (Sigma-Aldrich) for myoblasts.
  • WT or DM1 cells are differentiated for 4 days. Then, medium was changed with fresh differentiation medium with peptide-PMO conjugates at a 1 , 2 ,5 10, 20 or 40 pM concentration. Cells were harvested for analysis 48h after treatment. Cell viability was quantified in after 2 days of transfection of peptide-PMOs at 40uM in human hepatocytes or at a 1 , 2 ,5 10, 20 or 40 pM concentration in human myoblasts using a fluorescent-based assay (Promega). RNA isolation, RT-PCR and qPCR analysis.
  • mice tissues prior to RNA extraction, muscles were disrupted in TriReagent (Sigma- Aldrich) using Fastprep system and Lysing Matrix D tubes (MP biomedicals).
  • TriReagent Sigma- Aldrich
  • Lysing Matrix D tubes MP biomedicals.
  • human cells prior to RNA extraction, cells were lysed in a proteinase K buffer (500 mM NaCI, 10 mM Tris- HCI, pH 7.2, 1.5 mM MgCI2, 10 mM EDTA, 2% SDS and 0.5 mg/ml of proteinase K) for 45 min at 55°C. Total RNAs were isolated using TriReagent according to the manufacturer’s protocol.
  • RNA was reverse transcribed using M-MLV first-strand synthesis system (Life Technologies) according to the manufacturer’s instructions in a total of 20 pL.
  • M-MLV first-strand synthesis system Life Technologies
  • One microliter of cDNA preparation was subsequently used in a semi-quantitative PCR analysis according to standard protocol (ReddyMix, Thermo Scientific). PCR amplification was carried out for 25-35 cycles within the linear range of amplification for each gene. PCR products were resolved on 1.5-2% agarose gels, ethidium bromide-stained and quantified with ImageJ software. The ratios of exon inclusion were quantified as a percentage of inclusion relative to total intensity of isoform signals. Primers are shown in the following table 7:
  • the treated DM1 patient derived muscle cells showed that the DPEP 1 or 3 peptide-[CAG] 7 PMO conjugates specifically target mutant CUGexp-DMPK transcripts to abrogate the detrimental sequestration of MBNL1 splicing factor by nuclear RNA foci and consequently MBNL1 functional loss, responsible for splicing defects and muscle dysfunction.
  • the DPEP1/3 peptide-[CAG] 7 PMO conjugates penetrate cells and induce splicing normalisation with high efficacy (figure 13).
  • conjugates formed with DPEP1/3 indicate that ALP, ALT, AST, KIM-1 , BUN, NGAL, and creatinine levels were similar to saline control injections, in contrast to the fold increases typically induced by currently available peptide carriers from the Pip series.
  • conjugates formed from DPEP peptides with a [CAG] 7 PMO are as active as conjugates formed with prior peptides such as Pip6a yet have wider therapeutic window because they are less toxic (figures 15-19).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des peptides, en particulier des peptides de pénétration cellulaire, et des conjugués de tels peptides de pénétration cellulaire avec une molécule thérapeutique. La présente invention concerne en outre l'utilisation de tels peptides ou conjugués dans des procédés de traitement ou en tant que médicament, en particulier dans le traitement de troubles génétiques et en particulier de dystrophies musculaires telles que la dystrophie musculaire de Duchenne.
PCT/GB2019/052247 2018-08-09 2019-08-09 Peptides de pénétration cellulaire WO2020030927A1 (fr)

Priority Applications (10)

Application Number Priority Date Filing Date Title
AU2019317789A AU2019317789A1 (en) 2018-08-09 2019-08-09 Cell-penetrating peptides
KR1020217007003A KR20210090157A (ko) 2018-08-09 2019-08-09 세포 투과 펩티드
CA3108876A CA3108876A1 (fr) 2018-08-09 2019-08-09 Peptides de penetration cellulaire
JP2021506986A JP2021534131A (ja) 2018-08-09 2019-08-09 細胞透過性ペプチド
EP19755420.7A EP3833376A1 (fr) 2018-08-09 2019-08-09 Peptides de pénétration cellulaire
MX2021001545A MX2021001545A (es) 2018-08-09 2019-08-09 Péptidos penetrantes celulares.
BR112021002447-6A BR112021002447A2 (pt) 2018-08-09 2019-08-09 peptídeos de penetração celular
CN201980066823.5A CN113347989A (zh) 2018-08-09 2019-08-09 细胞穿透肽
US17/266,939 US20210299263A1 (en) 2018-08-09 2019-08-09 Cell-penetrating peptides
IL280678A IL280678A (en) 2018-08-09 2021-02-07 Cell-penetrating peptides

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1812972.6A GB201812972D0 (en) 2018-08-09 2018-08-09 Cell-penetrating peptides
GB1812972.6 2018-08-09

Publications (1)

Publication Number Publication Date
WO2020030927A1 true WO2020030927A1 (fr) 2020-02-13

Family

ID=63667311

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2019/052247 WO2020030927A1 (fr) 2018-08-09 2019-08-09 Peptides de pénétration cellulaire

Country Status (12)

Country Link
US (1) US20210299263A1 (fr)
EP (1) EP3833376A1 (fr)
JP (1) JP2021534131A (fr)
KR (1) KR20210090157A (fr)
CN (1) CN113347989A (fr)
AU (1) AU2019317789A1 (fr)
BR (1) BR112021002447A2 (fr)
CA (1) CA3108876A1 (fr)
GB (1) GB201812972D0 (fr)
IL (1) IL280678A (fr)
MX (1) MX2021001545A (fr)
WO (1) WO2020030927A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022172019A1 (fr) 2021-02-12 2022-08-18 Oxford University Innovation Limited Conjugués peptidiques à pénétration cellulaire et leurs procédés d'utilisation
WO2022192749A3 (fr) * 2021-03-12 2022-10-27 Pepgen Inc. Méthodes de traitement de la dystrophie musculaire de duchenne à l'aide de conjugués peptide-oligonucléotide
WO2022271818A1 (fr) * 2021-06-23 2022-12-29 Entrada Therapeutics, Inc. Composés antisens et méthodes de ciblage de répétitions de cug
US11987647B2 (en) 2018-05-09 2024-05-21 Ohio State Innovation Foundation Cyclic cell-penetrating peptides with one or more hydrophobic residues

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009147368A1 (fr) * 2008-06-04 2009-12-10 Medical Research Council Peptides
EP2394665A1 (fr) * 2010-06-14 2011-12-14 F. Hoffmann-La Roche AG Peptides pénétrant dans la cellule et leurs utilisations
US20120289457A1 (en) * 2011-05-05 2012-11-15 Avi Biopharma, Inc. Peptide oligonucleotide conjugates
WO2013030569A2 (fr) * 2011-08-30 2013-03-07 Michael John Gait Peptides
WO2015022504A2 (fr) * 2013-08-12 2015-02-19 Medical Research Council Conjugués peptidiques
WO2016028187A1 (fr) 2014-08-22 2016-02-25 Noogen Llc Oligonucléotides modifiés et leurs procédés de synthèse

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2011267078B2 (en) * 2010-06-14 2014-09-25 F. Hoffmann-La Roche Ag Cell-penetrating peptides and uses therof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009147368A1 (fr) * 2008-06-04 2009-12-10 Medical Research Council Peptides
EP2394665A1 (fr) * 2010-06-14 2011-12-14 F. Hoffmann-La Roche AG Peptides pénétrant dans la cellule et leurs utilisations
US20120289457A1 (en) * 2011-05-05 2012-11-15 Avi Biopharma, Inc. Peptide oligonucleotide conjugates
WO2013030569A2 (fr) * 2011-08-30 2013-03-07 Michael John Gait Peptides
WO2015022504A2 (fr) * 2013-08-12 2015-02-19 Medical Research Council Conjugués peptidiques
WO2016028187A1 (fr) 2014-08-22 2016-02-25 Noogen Llc Oligonucléotides modifiés et leurs procédés de synthèse

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 2000, LIPPINCOTT, WILLIAMS & WILKINS
ABLAN FRANCIS D O ET AL: "Charge Distribution Fine-Tunes the Translocation of[alpha]-Helical Amphipathic Peptides across Membranes", BIOPHYSICAL JOURNAL, ELSEVIER, AMSTERDAM, NL, vol. 111, no. 8, 18 October 2016 (2016-10-18), pages 1738 - 1749, XP029771875, ISSN: 0006-3495, DOI: 10.1016/J.BPJ.2016.08.047 *
BAHAL, NAT.COMM., 2016
CORINNE BETTS ET AL: "Pip6-PMO, A New Generation of Peptide-oligonucleotide Conjugates With Improved Cardiac Exon Skipping Activity for DMD Treatment", MOLECULAR THERAPY - NUCLEIC ACIDS, vol. 1, no. 8, 1 August 2012 (2012-08-01), pages e38, XP055128893, ISSN: 2162-2531, DOI: 10.1038/mtna.2012.30 *
FRANCESCA MILLETTI: "Cell-penetrating peptides: classes, origin, and current landscape", DRUG DISCOVERY TODAY, vol. 17, no. 15-16, 1 August 2012 (2012-08-01), pages 850 - 860, XP055102101, ISSN: 1359-6446, DOI: 10.1016/j.drudis.2012.03.002 *
HANNA A. RYDBERG ET AL: "Effects of Tryptophan Content and Backbone Spacing on the Uptake Efficiency of Cell-Penetrating Peptides", BIOCHEMISTRY, vol. 51, no. 27, 28 June 2012 (2012-06-28), pages 5531 - 5539, XP055612466, ISSN: 0006-2960, DOI: 10.1021/bi300454k *
LUKASZ KOZLOWSKI, BIOL DIRECT, vol. 11, 2016, pages 55
OSMAN ET AL., HMG, 2014
R. P. WU ET AL: "Cell-penetrating peptides as transporters for morpholino oligomers: effects of amino acid composition on intracellular delivery and cytotoxicity", NUCLEIC ACIDS RESEARCH, vol. 35, no. 15, 1 January 2007 (2007-01-01), pages 5182 - 5191, XP055049876, ISSN: 0305-1048, DOI: 10.1093/nar/gkm478 *
SAMBROOK, J.RUSSELL, D.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS
W.C. WIMLEYS.H. WHITE: "Experimentally determined hydrophobicity scale for proteins at membrane interfaces", NATURE STRUCT BIOL, vol. 3, 1996, pages 842, XP000938728, doi:10.1038/nsb1096-842
ZHOU, HGT, 2013

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11987647B2 (en) 2018-05-09 2024-05-21 Ohio State Innovation Foundation Cyclic cell-penetrating peptides with one or more hydrophobic residues
WO2022172019A1 (fr) 2021-02-12 2022-08-18 Oxford University Innovation Limited Conjugués peptidiques à pénétration cellulaire et leurs procédés d'utilisation
WO2022171972A1 (fr) 2021-02-12 2022-08-18 Oxford University Innovation Limited Conjugués peptidiques de pénétration cellulaire et leurs procédés d'utilisation
WO2022192749A3 (fr) * 2021-03-12 2022-10-27 Pepgen Inc. Méthodes de traitement de la dystrophie musculaire de duchenne à l'aide de conjugués peptide-oligonucléotide
WO2022271818A1 (fr) * 2021-06-23 2022-12-29 Entrada Therapeutics, Inc. Composés antisens et méthodes de ciblage de répétitions de cug

Also Published As

Publication number Publication date
US20210299263A1 (en) 2021-09-30
EP3833376A1 (fr) 2021-06-16
KR20210090157A (ko) 2021-07-19
JP2021534131A (ja) 2021-12-09
CN113347989A (zh) 2021-09-03
GB201812972D0 (en) 2018-09-26
AU2019317789A1 (en) 2021-03-04
IL280678A (en) 2021-03-25
BR112021002447A2 (pt) 2021-05-04
MX2021001545A (es) 2021-08-24
CA3108876A1 (fr) 2020-02-13

Similar Documents

Publication Publication Date Title
US20210299263A1 (en) Cell-penetrating peptides
CA2846218C (fr) Peptides penetrant la cellule comportant un domaine hydrophobe central
JP2011523557A (ja) ペプチド
US20220125934A1 (en) Linkers
US20220275372A1 (en) Conjugate and uses thereof
US20220041662A1 (en) Cell penetrating peptides
US20210299264A1 (en) Cell-penetrating peptides
WO2023194729A1 (fr) Peptides de pénétration cellulaire riches en lysine
WO2024094980A1 (fr) Peptides de pénétration cellulaire raccourcis
WO2022172019A1 (fr) Conjugués peptidiques à pénétration cellulaire et leurs procédés d'utilisation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19755420

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3108876

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021506986

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021002447

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019317789

Country of ref document: AU

Date of ref document: 20190809

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019755420

Country of ref document: EP

Effective date: 20210309

ENP Entry into the national phase

Ref document number: 112021002447

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210209